1
|
Fiorentino G, Merico V, Zanoni M, Comincini S, Sproviero D, Garofalo M, Gagliardi S, Cereda C, Lin CJ, Innocenti F, Taggi M, Vaiarelli A, Ubaldi FM, Rienzi L, Cimadomo D, Garagna S, Zuccotti M. Extracellular vesicles secreted by cumulus cells contain microRNAs that are potential regulatory factors of mouse oocyte developmental competence. Mol Hum Reprod 2024; 30:gaae019. [PMID: 38745364 DOI: 10.1093/molehr/gaae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
The role of cumulus cells (CCs) in the acquisition of oocyte developmental competence is not yet fully understood. In a previous study, we matured cumulus-denuded fully-grown mouse oocytes to metaphase II (MII) on a feeder layer of CCs (FL-CCs) isolated from developmentally competent (FL-SN-CCs) or incompetent (FL-NSN-CCs) SN (surrounded nucleolus) or NSN (not surrounding nucleolus) oocytes, respectively. We observed that oocytes cultured on the former could develop into blastocysts, while those matured on the latter arrested at the 2-cell stage. To investigate the CC factors contributing to oocyte developmental competence, here we focused on the CCs' release into the medium of extracellular vesicles (EVs) and on their miRNA content. We found that, during the 15-h transition to MII, both FL-SN-CCs and FL-NSN-CCs release EVs that can be detected, by confocal microscopy, inside the zona pellucida (ZP) or the ooplasm. The majority of EVs are <200 nm in size, which is compatible with their ability to cross the ZP. Next-generation sequencing of the miRNome of FL-SN-CC versus FL-NSN-CC EVs highlighted 74 differentially expressed miRNAs, with 43 up- and 31 down-regulated. Although most of these miRNAs do not have known roles in the ovary, in silico functional analysis showed that seven of these miRNAs regulate 71 target genes with specific roles in meiosis resumption (N = 24), follicle growth (N = 23), fertilization (N = 1), and the acquisition of oocyte developmental competence (N = 23). Overall, our results indicate CC EVs as emerging candidates of the CC-to-oocyte communication axis and uncover a group of miRNAs as potential regulatory factors.
Collapse
Affiliation(s)
- Giulia Fiorentino
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology 'Lazzaro Spallanzani', University of Pavia, Pavia, Italy
| | - Valeria Merico
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology 'Lazzaro Spallanzani', University of Pavia, Pavia, Italy
| | - Mario Zanoni
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology 'Lazzaro Spallanzani', University of Pavia, Pavia, Italy
| | - Sergio Comincini
- Functional Genomics Laboratory, Department of Biology and Biotechnology 'Lazzaro Spallanzani', University of Pavia, Pavia, Italy
| | - Daisy Sproviero
- IFOM, IFOM-The FIRC Institute of Molecular Oncology, Milan, Italy
| | - Maria Garofalo
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Stella Gagliardi
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Cristina Cereda
- Department of Pediatrics, Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, Milan, Italy
| | - Chih-Jen Lin
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Federica Innocenti
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Marilena Taggi
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Alberto Vaiarelli
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | | | - Laura Rienzi
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Danilo Cimadomo
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Silvia Garagna
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology 'Lazzaro Spallanzani', University of Pavia, Pavia, Italy
| | - Maurizio Zuccotti
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology 'Lazzaro Spallanzani', University of Pavia, Pavia, Italy
| |
Collapse
|
2
|
Wijesena HR, Keel BN, Nonneman DJ, Cushman RA, Lents CA. Clustering of multi-tissue transcriptomes in gilts with normal cyclicity or delayed puberty reveals genes related to pubertal development†. Biol Reprod 2024; 110:261-274. [PMID: 37870496 DOI: 10.1093/biolre/ioad145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023] Open
Abstract
In gilts, puberty is marked by standing estrus in the presence of a boar. Delayed puberty (DP; failure to display pubertal estrus) is a major reason for gilt removal. To investigate the physiological determinants underlying DP in gilts, transcriptomic data from tissues relevant to estrus and puberty, such as mediobasal hypothalamus, anterior pituitary gland, ovarian cortex, olfactory bulb, amygdala, and hippocampus, were obtained from age-matched DP (n = 8) and cyclic control gilts at follicular phase (n = 8) and luteal phase (n = 8) of the estrous cycle. A gene expression module analysis via three-way gene × individual × tissue clustering using tensor decomposition identified pituitary and ovary gene modules contributing to regulation of pubertal development. Analysis of gene expression in the hypothalamic-pituitary-ovary axis identified reduced expression of hypothalamic genes critical for stimulating gonadotropin secretion (KISS1 and TAC3) and reduced expression of LHB in the anterior pituitary of DP gilts compared with their cyclic counterparts. Consequently, luteinizing hormone-induced genes in the ovary important for folliculogenesis (OXTR, RUNX2, and PTX3) were less expressed in DP gilts. Other intrafollicular genes (AHR, PTGS2, PTGFR, and IGFBP7) and genes in the steroidogenesis pathways (STAR and CYP11A1) necessary to complete the ovulatory cascade were also less expressed in DP gilts. This is the first clustering of multi-tissue expression data from DP and cyclic gilts to identify genes differentially expressed in gilts of similar ages but at different levels of sexual development. A critical lack of gonadotropin support and reduced ovarian responsiveness underlie DP in gilts.
Collapse
Affiliation(s)
| | - Brittney N Keel
- USDA, ARS, US Meat Animal Research Center, Clay Center, NE, USA
| | - Dan J Nonneman
- USDA, ARS, US Meat Animal Research Center, Clay Center, NE, USA
| | | | - Clay A Lents
- USDA, ARS, US Meat Animal Research Center, Clay Center, NE, USA
| |
Collapse
|
3
|
Mantri M, Zhang HH, Spanos E, Ren YA, De Vlaminck I. A spatiotemporal molecular atlas of the ovulating mouse ovary. Proc Natl Acad Sci U S A 2024; 121:e2317418121. [PMID: 38252830 PMCID: PMC10835069 DOI: 10.1073/pnas.2317418121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Ovulation is essential for reproductive success, yet the underlying cellular and molecular mechanisms are far from clear. Here, we applied high-resolution spatiotemporal transcriptomics to map out cell type- and ovulation stage-specific molecular programs as function of time during follicle maturation and ovulation in mice. Our analysis revealed dynamic molecular transitions within granulosa cell types that occur in tight coordination with mesenchymal cell proliferation. We identified molecular markers for the emerging cumulus cell fate during the preantral-to-antral transition. We describe transcriptional programs that respond rapidly to ovulation stimulation and those associated with follicle rupture, highlighting the prominent roles of apoptotic and metabolic pathways during the final stages of follicle maturation. We further report stage-specific oocyte-cumulus cell interactions and diverging molecular differentiation in follicles approaching ovulation. Collectively, this study provides insights into the cellular and molecular processes that regulate mouse ovarian follicle maturation and ovulation with important implications for advancing therapeutic strategies in reproductive medicine.
Collapse
Affiliation(s)
- Madhav Mantri
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY14850
| | | | - Emmanuel Spanos
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY14850
| | - Yi A. Ren
- Department of Animal Science, Cornell University, Ithaca, NY14850
| | - Iwijn De Vlaminck
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY14850
| |
Collapse
|
4
|
Medeiros SFD, Barbosa BB, Medeiros MASD, Yamamoto MMW. Morphology and Biochemistry of Ovulation. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2021; 43:480-486. [PMID: 34318473 PMCID: PMC10411198 DOI: 10.1055/s-0041-1731379] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/19/2021] [Indexed: 10/20/2022] Open
Abstract
The process of ovulation involves multiple and iterrelated genetic, biochemical, and morphological events: cessation of the proliferation of granulosa cells, resumption of oocyte meiosis, expansion of cumulus cell-oocyte complexes, digestion of the follicle wall, and extrusion of the metaphase-II oocyte. The present narrative review examines these interrelated steps in detail. The combined or isolated roles of the follicle-stimulating hormone (FSH) and luteinizing hormone (LH) are highlighted. Genes indiced by the FSH genes are relevant in the cumulus expansion, and LH-induced genes are critical for the resumption of meiosis and digestion of the follicle wall. A non-human model for follicle-wall digestion and oocyte release was provided.
Collapse
Affiliation(s)
- Sebastião Freitas de Medeiros
- Department of Gynecology and Obstetrics, Faculdade de Medicina, Universidade Federal do Mato Grosso, Cuiabá, Mato Grosso, MT, Brazil
- Instituto Tropical de Medicina Reprodutiva, Cuiabá, Mato Grosso, MT, Brazil
| | | | | | | |
Collapse
|
5
|
Yuan B, Yang J, Dubeau L, Hu Y, Li R. A Phosphotyrosine Switch in Estrogen Receptor β Is Required for Mouse Ovarian Function. Front Cell Dev Biol 2021; 9:649087. [PMID: 33898441 PMCID: PMC8063698 DOI: 10.3389/fcell.2021.649087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/16/2021] [Indexed: 11/13/2022] Open
Abstract
The two homologous estrogen receptors ERα and ERβ exert distinct effects on their cognate tissues. Previous work from our laboratory identified an ERβ-specific phosphotyrosine residue that regulates ERβ transcriptional activity and antitumor function in breast cancer cells. To determine the physiological role of the ERβ phosphotyrosine residue in normal tissue development and function, we investigated a mutant mouse model (Y55F) whereby this particular tyrosine residue in endogenous mouse ERβ is mutated to phenylalanine. While grossly indistinguishable from their wild-type littermates, mutant female mice displayed reduced fertility, decreased ovarian follicular cell proliferation, and lower progesterone levels. Moreover, mutant ERβ from female mice during superovulation is defective in activating promoters of its target genes in ovarian tissues. Thus, our findings provide compelling genetic and molecular evidence for a role of isotype-specific ERβ phosphorylation in mouse ovarian development and function.
Collapse
Affiliation(s)
- Bin Yuan
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Jing Yang
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Louis Dubeau
- Department of Pathology, USC/Norris Comprehensive Cancer Center, Keck School of Medicine of University of Southern California, Los Angeles, CA, United States
| | - Yanfen Hu
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Rong Li
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| |
Collapse
|
6
|
Lee-Thacker S, Jeon H, Choi Y, Taniuchi I, Takarada T, Yoneda Y, Ko C, Jo M. Core Binding Factors are essential for ovulation, luteinization, and female fertility in mice. Sci Rep 2020; 10:9921. [PMID: 32555437 PMCID: PMC7303197 DOI: 10.1038/s41598-020-64257-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/12/2020] [Indexed: 12/17/2022] Open
Abstract
Core Binding Factors (CBFs) are a small group of heterodimeric transcription factor complexes composed of DNA binding proteins, RUNXs, and a non-DNA binding protein, CBFB. The LH surge increases the expression of Runx1 and Runx2 in ovulatory follicles, while Cbfb is constitutively expressed. To investigate the physiological significance of CBFs, we generated a conditional mutant mouse model in which granulosa cell expression of Runx2 and Cbfb was deleted by the Esr2Cre. Female Cbfbflox/flox;Esr2cre/+;Runx2flox/flox mice were infertile; follicles developed to the preovulatory follicle stage but failed to ovulate. RNA-seq analysis of mutant mouse ovaries collected at 11 h post-hCG unveiled numerous CBFs-downstream genes that are associated with inflammation, matrix remodeling, wnt signaling, and steroid metabolism. Mutant mice also failed to develop corpora lutea, as evident by the lack of luteal marker gene expression, marked reduction of vascularization, and excessive apoptotic staining in unruptured poorly luteinized follicles, consistent with dramatic reduction of progesterone by 24 h after hCG administration. The present study provides in vivo evidence that CBFs act as essential transcriptional regulators of both ovulation and luteinization by regulating the expression of key genes that are involved in inflammation, matrix remodeling, cell differentiation, vascularization, and steroid metabolisms in mice.
Collapse
Affiliation(s)
- Somang Lee-Thacker
- Department of Obstetrics and Gynecology, Chandler Medical Center, 800 Rose Street, University of Kentucky, Lexington, KY, 40536-0298, USA
| | - Hayce Jeon
- Department of Obstetrics and Gynecology, Chandler Medical Center, 800 Rose Street, University of Kentucky, Lexington, KY, 40536-0298, USA
| | - Yohan Choi
- Department of Obstetrics and Gynecology, Chandler Medical Center, 800 Rose Street, University of Kentucky, Lexington, KY, 40536-0298, USA
| | - Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Takeshi Takarada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Yukio Yoneda
- Section of Prophylactic Pharmacology, Kanazawa University, Venture Business Laboratory 402, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - CheMyong Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, 2001 South Lincoln Avenue, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61802, USA
| | - Misung Jo
- Department of Obstetrics and Gynecology, Chandler Medical Center, 800 Rose Street, University of Kentucky, Lexington, KY, 40536-0298, USA.
| |
Collapse
|
7
|
Kannan S, Bhaskaran RS. Sustained obesity reduces litter size by decreasing proteins regulating folliculogenesis and ovulation in rats - A cafeteria diet model. Biochem Biophys Res Commun 2019; 519:475-480. [DOI: 10.1016/j.bbrc.2019.09.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/08/2019] [Indexed: 12/13/2022]
|
8
|
Nynca A, Sadowska A, Paukszto L, Molcan T, Ruszkowska M, Swigonska S, Orlowska K, Myszczynski K, Jastrzebski JP, Ciereszko RE. Temporal changes in the transcriptomic profile of granulosa cells of pigs treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Anim Reprod Sci 2019; 207:83-94. [PMID: 31213330 DOI: 10.1016/j.anireprosci.2019.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/15/2019] [Accepted: 06/04/2019] [Indexed: 01/17/2023]
Abstract
The 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) compound is an environmental chemical adversely affecting reproductive processes. Intracellular TCDD effects are mediated via aryl hydrocarbon receptor (AhR). The aim of the current study was to identify genes linking the AhR pathway with phenotypic consequences of TCDD action in granulosa cells of pigs. By applying multifactorial analysis, with TCDD and incubation time as factors, it was possible to determine temporal changes induced by TCDD in the cell transcriptome. Among the identified 144 differentially expressed genes (DEGs; Padjusted<0.05, log2 fold change (FC)≥1), 111 DEGs were classified as sustained genes (FC values changing between 3 and 24 h). Eighty six DEGs were classified as early genes and only nine as late genes (FC changes observed between 3 and 12 h or 12 and 24 h, respectively). The sustained gene category included genes related to TCDD mechanism of action (AHR, ARNTL, CYP1A1), cell proliferation (TGFβ3), follicular development and ovulation (PTGS2) as well as stress response (NR3C1). The early gene category contained DEGs associated with cell proliferation (DUSP4, TAB1) and cellular response to stress (DHX34). The CYP1A1 gene was the only DEG classified as an early, late and sustained gene. The multifactorial approach allowed for statistically analyzing TCDD-induced changes over time in the gene expression in granulosa cells of pigs. Changes over time in the granulosal transcriptome profile indicated the involvement of stress related molecules in the cellular response to TCDD and TCDD effects on ovulation. The TCDD effects were particularly evident during the early stage of action by this compound.
Collapse
Affiliation(s)
- Anna Nynca
- Laboratory of Molecular Diagnostics, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Prawochenskiego 5, 10-720, Olsztyn, Poland.
| | - Agnieszka Sadowska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719, Olsztyn, Poland.
| | - Lukasz Paukszto
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719, Olsztyn, Poland.
| | - Tomasz Molcan
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719, Olsztyn, Poland.
| | - Monika Ruszkowska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719, Olsztyn, Poland.
| | - Sylwia Swigonska
- Laboratory of Molecular Diagnostics, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Prawochenskiego 5, 10-720, Olsztyn, Poland.
| | - Karina Orlowska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719, Olsztyn, Poland.
| | - Kamil Myszczynski
- Department of Botany and Nature Protection, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Plac Lodzki 1, 10-727, Olsztyn, Poland.
| | - Jan P Jastrzebski
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719, Olsztyn, Poland.
| | - Renata E Ciereszko
- Laboratory of Molecular Diagnostics, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Prawochenskiego 5, 10-720, Olsztyn, Poland; Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719, Olsztyn, Poland.
| |
Collapse
|
9
|
Velásquez A, Mellisho E, Castro FO, Rodríguez-Álvarez L. Effect of BMP15 and/or AMH during in vitro maturation of oocytes from involuntarily culled dairy cows. Mol Reprod Dev 2018; 86:209-223. [PMID: 30548943 DOI: 10.1002/mrd.23096] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 12/03/2018] [Indexed: 01/10/2023]
Abstract
The high metabolic activity to which the dairy cattle are exposed to maintain milk production altered steroid metabolism that affects reproductive physiology and reduce oocyte competence. Our aims were (a) to characterize the competence of immature oocytes collected from dairy cattle based on the expression of genes in cumulus cells (CCs) and (b) to improve oocyte competence to support preimplantation embryo development by the supplementation of maturation medium with bone morphogenetic protein 15 (BMP15) and/or anti-mullerian hormone (AMH). Oocyte donors were identified at the moment of ovary collection and grouped by involuntarily culled dairy cows (Holstein breed) or beef cattle. The embryo development speed to blastocyst of the cull dairy cattle versus beef cattle (control group) was lower. Besides, <10% of oocytes (with CC biopsies) derived from dairy cattle were able to develop to the blastocyst stage. In addition, a higher level of expression and a positive correlation were observed in the expression of most of the genes evaluated (LUM, KRT18, KRT8, CLIC3, BMPR1B, and SLC38A3) in the cumulus-oocyte complexes that produced blastocysts versus those which did not develop correctly (arrested development). Further, use of BMP15 in the maturation of oocytes from dairy cattle seems to increase competence, modulating the expression of OCT4, SOX2, CDX2, GATA6, and TP1 in resulting blastocysts.
Collapse
Affiliation(s)
- Alejandra Velásquez
- Laboratory of Animal Biotechnology, Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile
| | - Edwin Mellisho
- Laboratory of Animal Biotechnology, Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile
| | - Fidel Ovidio Castro
- Laboratory of Animal Biotechnology, Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile
| | - Lleretny Rodríguez-Álvarez
- Laboratory of Animal Biotechnology, Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile
| |
Collapse
|
10
|
Tang H, Wang L, Chen Y, He J, Qu L, Guo Y, Liu Y, Liu X, Lin H. Ovulation is associated with the LH-dependent induction of pla2g4aa in zebrafish. Mol Cell Endocrinol 2018; 473:53-60. [PMID: 29326060 DOI: 10.1016/j.mce.2018.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/17/2017] [Accepted: 01/07/2018] [Indexed: 01/14/2023]
Abstract
The effects of the preovulatory luteinizing hormone (LH) surge on the ovulatory process are mediated by prostaglandins (PGs), the synthesis of which involves prostaglandin synthetase and cytosolic phospholipase A2 (cPLA2). In our previous study, we systematically investigated the function of prostaglandin endoperoxide synthase (ptgs) genes on ovulation in zebrafish. However, the role of cPLA2 in ovulation was not determined in zebrafish. In this study, we investigated the function of cpla2α in PGs production and ovulation in periovulatory follicles. Our data showed that the expression of pla2g4aa increased during zebrafish folliculogenesis and the follicular layer was the primary region with expression of pla2g4aa. In addition, the expression of pla2g4aa was regulated by LH in vitro and in vivo. Furthermore, injection of AACOCF3, a specific inhibitor of cPLA2, significantly reduced ovarian PGs level and blocked hCG-induced ovulation. Collectively, these findings suggest that pla2g4aa is related to the ovulation process in zebrafish.
Collapse
Affiliation(s)
- Haipei Tang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Le Wang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yu Chen
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Jianan He
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Ling Qu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yin Guo
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yun Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Xiaochun Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Haoran Lin
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| |
Collapse
|
11
|
Zhang P, Wang J, Lang H, Wang W, Liu X, Liu H, Tan C, Li X, Zhao Y, Wu X. Knockdown of CREB1 promotes apoptosis and decreases estradiol synthesis in mouse granulosa cells. Biomed Pharmacother 2018; 105:1141-1146. [PMID: 30021350 DOI: 10.1016/j.biopha.2018.06.101] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/18/2018] [Accepted: 06/18/2018] [Indexed: 12/17/2022] Open
Abstract
Cyclic AMP response element-binding protein 1 (CREB1), a member of the CREB family, is known to be involved in follicular growth, ovulation, and ovarian disease. However, the physiological function of CREB1 in mouse granulosa cells (mGCs) remains lagerly unknown. The aim of this study was to determine the role of CREB1 in mGCs by knocking down CREB1 expression. CREB1 knock-down in mGCs at the mRNA and protein levels, was confirmed by quantitative real-time polymerase chain reaction and western blot. Results of enzyme linked immunosorbent assay revealed that CREB1 knockdown significantly decreased the concentrations of estradiol (E2) and progesterone (P4) in mGCs. Furthermore, the CREB1 knockdown in mGCs promoted cell proliferation and apoptosis, and arrested the cell cycle in S-phase. To elucidate the regulatory mechanism underlying the effects of CREB1 knockdown on steroid synthesis, cell cycle, and apoptosis, we measured the protein expression levels of several related genes in mGCs knocked down CREB1. When CREB1 was knocked down, the expression of Cyp1b1 and Cyp19a1, which encode steroidogenic enzymes, was down-regulated; the expression of the cell cycle factors CyclinA1, CyclinB1, and CyclinD2 were significantly decreased. Among apoptosis-related genes, Bcl-2 was down-regulated, whereas Bax and cleaved Caspase3 were upregulated. Moreover, CREB1 knockdown significantly decreased expression level of Has2, Ptgs2, and Igfbp4, which are essential genes for folliculogenesis in mGCs. Taken together, these findings suggested that CREB1 might be a key regulator of mGCs through regulating steroid synthesis, cell proliferation, cell cycle, apoptosis, and other regulators of folliculogenesis.
Collapse
Affiliation(s)
- Pengju Zhang
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, #1363 Shengtai Street, Changchun 130124, Jilin Province, PR China
| | - Jun Wang
- College of Animal Science and Technology, Jilin Agricultural University, 2888 Xincheng Street, Changchun 130118, Jilin Province, PR China
| | - Hongyan Lang
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, #1363 Shengtai Street, Changchun 130124, Jilin Province, PR China
| | - Weixia Wang
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, #1363 Shengtai Street, Changchun 130124, Jilin Province, PR China
| | - Xiaohui Liu
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, #1363 Shengtai Street, Changchun 130124, Jilin Province, PR China
| | - Haiyan Liu
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, #1363 Shengtai Street, Changchun 130124, Jilin Province, PR China
| | - Chengcheng Tan
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, #1363 Shengtai Street, Changchun 130124, Jilin Province, PR China
| | - Xintao Li
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, #1363 Shengtai Street, Changchun 130124, Jilin Province, PR China.
| | - Yumin Zhao
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, #1363 Shengtai Street, Changchun 130124, Jilin Province, PR China.
| | - Xinghong Wu
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, #1363 Shengtai Street, Changchun 130124, Jilin Province, PR China.
| |
Collapse
|
12
|
Salustri A, Campagnolo L, Klinger FG, Camaioni A. Molecular organization and mechanical properties of the hyaluronan matrix surrounding the mammalian oocyte. Matrix Biol 2018; 78-79:11-23. [PMID: 29408277 DOI: 10.1016/j.matbio.2018.02.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 10/18/2022]
Abstract
Successful ovulation and oocyte fertilization are essential prerequisites for the beginning of life in sexually reproducing animals. In mammalian fertilization, the relevance of the protein coat surrounding the oocyte plasma membrane, known as zona pellucida, has been widely recognized, while, until not too long ago, the general belief was that the cumulus oophorus, consisting of follicle cells embedded in a hyaluronan rich extracellular matrix, was not essential. This opinion was based on in vitro fertilization procedures, in which a large number of sperms are normally utilized and the oocyte can be fertilized even if depleted of cumulus cells. Conversely, in vivo, only very few sperm cells reach the fertilization site, arguing against the possibility of a coincidental encounter with the oocyte. In the last two decades, proteins required for HA organization in the cumulus extracellular matrix have been identified and the study of fertility in mice deprived of the corresponding genes have provided compelling evidence that this jelly-like coat is critical for fertilization. This review focuses on the advances in understanding the molecular interactions making the cumulus environment suitable for oocyte and sperm encounter. Most of the studies on the molecular characterization of the cumulus extracellular matrix have been performed in the mouse and we will refer essentially to findings obtained in this animal model.
Collapse
Affiliation(s)
- Antonietta Salustri
- Department of Biomedicine and Prevention, Histology and Embryology Section, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Luisa Campagnolo
- Department of Biomedicine and Prevention, Histology and Embryology Section, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Francesca Gioia Klinger
- Department of Biomedicine and Prevention, Histology and Embryology Section, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Antonella Camaioni
- Department of Biomedicine and Prevention, Histology and Embryology Section, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
13
|
Tanaka Y, Kuwahara A, Ushigoe K, Yano Y, Taniguchi Y, Yamamoto Y, Matsuzaki T, Yasui T, Irahara M. Expression of cytokine-induced neutrophil chemoattractant suppresses tumor necrosis factor alpha expression and thereby prevents the follicles from undergoing atresia and apoptosis. Reprod Med Biol 2017; 16:157-165. [PMID: 29259464 PMCID: PMC5661809 DOI: 10.1002/rmb2.12022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/04/2017] [Indexed: 11/06/2022] Open
Abstract
Aim Cytokine-induced neutrophil chemoattractant (CINC/gro) is a CXC family chemokine, similar to interleukin-8 in rats, and is one of the factors that regulates ovulation. However, the mechanism that regulates atresia of the ovaries postovulation is not clearly defined. Methods Whether antibody-blocking of CINC/gro can alter the number of ovulated oocytes and modulate neutrophil infiltration was investigated. The effect of the antibody on the level of inflammatory cytokine production and follicular atresia was examined. Apoptosis was measured by the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) method and via analysis of the messenger RNA expression of Bcl-2 and Bcl2-associated X (Bax). Results The anti-CINC/gro antibody treatment decreased the number of ovulated oocytes. The messenger RNA levels of cyclooxygenase-2 and interleukin-1 beta were decreased by the antibody treatment, whereas that of tumor necrosis factor (TNF) alpha was increased. The TUNEL analysis revealed a larger number of apoptotic cells in the antibody group, compared with those in the control group, as well as a significant increase in the Bax/Bcl-2 ratio 24 hours after human chorionic gonadotropin administration. Conclusion These findings suggest that ovulation is accelerated by neutrophil infiltration into the theca layer. The CINC/gro appears to synergize with interleukin-1 beta for ovulation. By contrast, the data suggest that CINC/gro expression suppresses TNF alpha expression and that CINC/gro expression therefore prevents the follicles from undergoing atresia and apoptosis.
Collapse
Affiliation(s)
- Yu Tanaka
- Department of Obstetrics and Gynecology Japanese Red Cross Kochi Hospital Kochi Japan
| | - Akira Kuwahara
- Department of Obstetrics and Gynecology Institute of Health Biosciences The University of Tokushima Graduate School Tokushima Japan
| | - Kenjiro Ushigoe
- Department of Obstetrics and Gynecology Institute of Health Biosciences The University of Tokushima Graduate School Tokushima Japan
| | - Yuya Yano
- Department of Obstetrics and Gynecology Institute of Health Biosciences The University of Tokushima Graduate School Tokushima Japan
| | - Yuka Taniguchi
- Department of Obstetrics and Gynecology Institute of Health Biosciences The University of Tokushima Graduate School Tokushima Japan
| | - Yuri Yamamoto
- Department of Obstetrics and Gynecology Institute of Health Biosciences The University of Tokushima Graduate School Tokushima Japan
| | - Toshiya Matsuzaki
- Department of Obstetrics and Gynecology Institute of Health Biosciences The University of Tokushima Graduate School Tokushima Japan
| | - Toshiyuki Yasui
- Department of Obstetrics and Gynecology Institute of Health Biosciences The University of Tokushima Graduate School Tokushima Japan
| | - Minoru Irahara
- Department of Obstetrics and Gynecology Institute of Health Biosciences The University of Tokushima Graduate School Tokushima Japan
| |
Collapse
|
14
|
Choi Y, Wilson K, Hannon PR, Rosewell KL, Brännström M, Akin JW, Curry TE, Jo M. Coordinated Regulation Among Progesterone, Prostaglandins, and EGF-Like Factors in Human Ovulatory Follicles. J Clin Endocrinol Metab 2017; 102:1971-1982. [PMID: 28323945 PMCID: PMC5470773 DOI: 10.1210/jc.2016-3153] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 03/06/2017] [Indexed: 01/12/2023]
Abstract
CONTEXT In animal models, the luteinizing hormone surge increases progesterone (P4) and progesterone receptor (PGR), prostaglandins (PTGs), and epidermal growth factor (EGF)-like factors that play essential roles in ovulation. However, little is known about the expression, regulation, and function of these key ovulatory mediators in humans. OBJECTIVE To determine when and how these key ovulatory mediators are induced after the luteinizing hormone surge in human ovaries. DESIGN AND PARTICIPANTS Timed periovulatory follicles were obtained from cycling women. Granulosa/lutein cells were collected from in vitro fertilization patients. MAIN OUTCOME MEASURES The in vivo and in vitro expression of PGR, PTG synthases and transporters, and EGF-like factors were examined at the level of messenger RNA and protein. PGR binding to specific genes was assessed. P4 and PTGs in conditioned media were measured. RESULTS PGR, PTGS2, and AREG expressions dramatically increased in ovulatory follicles at 12 to 18 hours after human chorionic gonadotropin (hCG). In human granulosa/lutein cell cultures, hCG increased P4 and PTG production and the expression of PGR, specific PTG synthases and transporters, and EGF-like factors, mimicking in vivo expression patterns. Inhibitors for P4/PGR and EGF-signaling pathways reduced hCG-induced increases in PTG production and the expression of EGF-like factors. PGR bound to the PTGS2, PTGES, and SLCO2A1 genes. CONCLUSIONS This report demonstrated the time-dependent induction of PGR, AREG, and PTGS2 in human periovulatory follicles. In vitro studies indicated that collaborative actions of P4/PGR and EGF signaling are required for hCG-induced increases in PTG production and potentiation of EGF signaling in human periovulatory granulosa cells.
Collapse
Affiliation(s)
- Yohan Choi
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Kalin Wilson
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Patrick R Hannon
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Katherine L Rosewell
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Mats Brännström
- Department of Obstetrics and Gynecology, University of Gothenburg, 405 30 Gothenburg, Sweden
- Stockholm IVF, 112 81 Stockholm, Sweden
| | - James W Akin
- Bluegrass Fertility Center, Lexington, Kentucky 40503
| | - Thomas E Curry
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Misung Jo
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| |
Collapse
|
15
|
Xiong Y, Chen H, Lin P, Wang A, Wang L, Jin Y. ATF6 knockdown decreases apoptosis, arrests the S phase of the cell cycle, and increases steroid hormone production in mouse granulosa cells. Am J Physiol Cell Physiol 2017; 312:C341-C353. [PMID: 28100484 DOI: 10.1152/ajpcell.00222.2016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/01/2016] [Accepted: 11/23/2016] [Indexed: 12/18/2022]
Abstract
Activating transcription factor 6 (ATF6), a sensor protein located in the endoplasmic reticulum (ER) membrane, is an important factor in the ER stress signaling pathway. ER stress is known to be involved in folliculogenesis, follicular growth, and ovulation; however, the physiological function of ATF6 in mouse granulosa cells remains largely unknown. The aim of this study was to assess the role of ATF6 in mouse granulosa cells with respect to apoptosis, the cell cycle, and steroid hormone production, as well as several key genes related to follicular development, via RNA interference, immunohistochemical staining, real-time quantitative PCR, Western blotting, flow cytometry, terminal deoxynucleotidyltransferase-mediated deoxy-UTP nick end labeling (TUNEL) assay, and ELISA. Immunohistochemical staining revealed that ATF6 was extensively distributed in the granulosa cells of various ovarian follicles and oocytes in adult female mice. FSH or LH treatment significantly increased ATF6 protein levels in mouse granulosa cells. In the meantime, a recombinant plasmid was used to deplete ATF6 successfully using short hairpin RNA-mediated interference technology, which was verified at both the mRNA and protein levels. Flow cytometry and TUNEL assay analysis indicated that ATF6 depletion decreased apoptosis and arrested the S phase of the cell cycle in mouse granulosa cells. Consistent with these results, p53, caspase-3, B cell lymphoma 2 (Bcl-2)-associated X protein, CCAAT-enhancer-binding protein homologous protein, cyclin A1, cyclin B1, and cyclin D2 mRNA expression decreased, whereas Bcl-2 and glucose-regulated protein 78 kDa mRNA expression increased. Interestingly, ATF6 knockdown obviously increased progesterone and estradiol production in mouse granulosa cells. Cytochrome P450 1b1 (Cyp1b1) mRNA levels were downregulated, whereas Cyp11a1, steroidogenic acute regulatory, and Cyp19a1 mRNA levels were upregulated, in keeping with the changes in steroid hormones. Furthermore, ATF6 disruption remarkably increased insulin-like growth factor binding protein4 (Igfbp4) expression and decreased hyaluronan synthase 2 (Has2), prostaglandin-endoperoxide synthase 2 (Ptgs2), and prostaglandin F receptor (Ptgfr) expression in mouse granulosa cells, which are proteins crucial for follicular development. But, after treating with tunicamycin, the levels of Has2, Ptgs2, and Ptgfr increased relatively, whereas Igfbp4 expression decreased. Collectively, these results imply that ATF6, as a key player in ER stress signaling, may regulate apoptosis, the cell cycle, steroid hormone synthesis, and other modulators related to folliculogenesis in mouse granulosa cells, which may indirectly be involved in the development, ovulation, and atresia of ovarian follicles by affecting the physiological function of granulosa cells. The present study extends our understanding and provides new insights into the physiological significance of ATF6, a key signal transducer of ER stress, in ovarian granulosa cells.
Collapse
Affiliation(s)
- Yongjie Xiong
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; and.,College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Huatao Chen
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; and.,College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Pengfei Lin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; and.,College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Aihua Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Lei Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; and.,College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yaping Jin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; and .,College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
16
|
Guo J, Shi L, Gong X, Jiang M, Yin Y, Zhang X, Yin H, Li H, Emori C, Sugiura K, Eppig JJ, Su YQ. Oocyte-dependent activation of MTOR in cumulus cells controls the development and survival of cumulus-oocyte complexes. J Cell Sci 2016; 129:3091-103. [PMID: 27358481 DOI: 10.1242/jcs.182642] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 06/28/2016] [Indexed: 01/12/2023] Open
Abstract
Communication between oocytes and their companion somatic cells promotes the healthy development of ovarian follicles, which is crucial for producing oocytes that can be fertilized and are competent to support embryogenesis. However, how oocyte-derived signaling regulates these essential processes remains largely undefined. Here, we demonstrate that oocyte-derived paracrine factors, particularly GDF9 and GDF9-BMP15 heterodimer, promote the development and survival of cumulus-cell-oocyte complexes (COCs), partly by suppressing the expression of Ddit4l, a negative regulator of MTOR, and enabling the activation of MTOR signaling in cumulus cells. Cumulus cells expressed less Ddit4l mRNA and protein than mural granulosa cells, which is in striking contrast to the expression of phosphorylated RPS6 (a major downstream effector of MTOR). Knockdown of Ddit4l activated MTOR signaling in cumulus cells, whereas inhibition of MTOR in COCs compromised oocyte developmental competence and cumulus cell survival, with the latter likely to be attributable to specific changes in a subset of transcripts in the transcriptome of COCs. Therefore, oocyte suppression of Ddit4l expression allows for MTOR activation in cumulus cells, and this oocyte-dependent activation of MTOR signaling in cumulus cells controls the development and survival of COCs.
Collapse
Affiliation(s)
- Jing Guo
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Lanying Shi
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Xuhong Gong
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Mengjie Jiang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Yaoxue Yin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Xiaoyun Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Hong Yin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Hui Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Chihiro Emori
- Department of Animal Resource Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Koji Sugiura
- Department of Animal Resource Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - John J Eppig
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - You-Qiang Su
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China Key Laboratory of Model Animal Research, Nanjing Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| |
Collapse
|
17
|
Shimada M, Umehara T, Hoshino Y. Roles of epidermal growth factor (EGF)-like factor in the ovulation process. Reprod Med Biol 2016; 15:201-216. [PMID: 29259438 DOI: 10.1007/s12522-016-0236-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/01/2016] [Indexed: 12/22/2022] Open
Abstract
Luteinizing hormone (LH) surge stimulates preovulatory follicles to induce the ovulation process, including oocyte maturation, cumulus expansion, and granulosa cell luteinization. The matured oocytes surrounded by an expanded cumulus cell layer are released from follicles to the oviduct. However, LH receptors are dominantly expressed in granulosa cells, but less in cumulus cells and are not expressed in oocytes, indicating that the secondary factors expressed and secreted from LH-stimulated granulosa cells are required for the induction of the ovulation process. Prostaglandin and progesterone are well-known factors that are produced in granulosa cells and then stimulate in both granulosa and cumulus cells. The mutant mice of prostaglandin synthase (Ptgs2KO mice) or progesterone receptor (PRKO mice) revealed that the functions were essential to accomplish the ovulation process, but not to induce the ovulation process. To identify the factors initiating the transfer of the stimuli of LH surge from granulosa cells to cumulus cells, M. Conti's lab and our group performed microarray analysis of granulosa cells and identified the epidermal growth factor (EGF)-like factor, amphiregulin (AREG), epiregulin (EREG), and β-cellulin (BTC) that act on EGF receptor (EGFR) and then induce the ERK1/2 and Ca2+-PLC pathways in cumulus cells. When each of the pathways was down-regulated using a pharmacological approach or gene targeting study, the induction of cumulus expansion and oocyte maturation were dramatically suppressed, indicating that both pathways are inducers of the ovulation process. However, an in vitro culture study also revealed that the EGFR-induced unphysiological activation of PKC in cumulus cells accelerated oocyte maturation with low cytostatic activity. Thus, the matured oocytes are not arrested at the metaphase II (MII) stage and then spontaneously form pronuclei. The expression of another type of EGF-like factor, neuregulin 1 (NRG1), that does not act on EGFR, but selectively binds to ErbB3 is observed in granulosa cells after the LH surge. NRG1 supports EGFR-induced ERK1/2 phosphorylation, but reduces PKC activity to physiological level in the cumulus cells, which delays the timing of meiotic maturation of oocytes to adjust the timing of ovulation. Thus, both types of EGF-like factor are rapidly induced by LH surge and then stimulate cumulus cells to control ERK1/2 and PKC pathways, which results in the release of matured oocytes with a fertilization competence.
Collapse
Affiliation(s)
- Masayuki Shimada
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science Hiroshima University Higashi-Hiroshima 739-8528 Hiroshima Japan
| | - Takashi Umehara
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science Hiroshima University Higashi-Hiroshima 739-8528 Hiroshima Japan
| | - Yumi Hoshino
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science Hiroshima University Higashi-Hiroshima 739-8528 Hiroshima Japan
| |
Collapse
|
18
|
El-Hayek S, Clarke HJ. Control of Oocyte Growth and Development by Intercellular Communication Within the Follicular Niche. Results Probl Cell Differ 2016; 58:191-224. [PMID: 27300180 DOI: 10.1007/978-3-319-31973-5_8] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In the mammalian ovary, each oocyte grows and develops within its own structural and developmental niche-the follicle. Together with the female germ cell in the follicle are somatic granulosa cells, specialized companion cells that surround the oocyte and provide support to it, and an outer layer of thecal cells that serve crucial roles including steroid synthesis. These follicular compartments function as a single physiological unit whose purpose is to produce a healthy egg, which upon ovulation can be fertilized and give rise to a healthy embryo, thus enabling the female germ cell to fulfill its reproductive potential. Beginning from the initial stage of follicle formation and until terminal differentiation at ovulation, oocyte and follicle growth depend absolutely on cooperation between the different cellular compartments. This cooperation synchronizes the initiation of oocyte growth with follicle activation. During growth, it enables metabolic support for the follicle-enclosed oocyte and allows the follicle to fulfill its steroidogenic potential. Near the end of the growth period, intra-follicular interactions prevent the precocious meiotic resumption of the oocyte and ensure its nuclear differentiation. Finally, cooperation enables the events of ovulation, including meiotic maturation of the oocyte and expansion of the cumulus granulosa cells. In this chapter, we discuss the cellular interactions that enable the growing follicle to produce a healthy oocyte, focusing on the communication between the germ cell and the surrounding granulosa cells.
Collapse
Affiliation(s)
- Stephany El-Hayek
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
- Department of Biology, McGill University, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Block E-M0.2218, Montreal, QC, Canada, H4A 3J1
| | - Hugh J Clarke
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada.
- Department of Biology, McGill University, Montreal, QC, Canada.
- Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Block E-M0.2218, Montreal, QC, Canada, H4A 3J1.
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
19
|
Bazzano M, Torelli C, Pustovrh M, Paz D, Elia E. Obesity induced by cafeteria diet disrupts fertility in the rat by affecting multiple ovarian targets. Reprod Biomed Online 2015; 31:655-67. [DOI: 10.1016/j.rbmo.2015.08.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 08/03/2015] [Accepted: 08/06/2015] [Indexed: 11/24/2022]
|
20
|
American ginseng regulates gene expression to protect against premature ovarian failure in rats. BIOMED RESEARCH INTERNATIONAL 2015; 2015:767124. [PMID: 25705687 PMCID: PMC4330957 DOI: 10.1155/2015/767124] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 11/30/2014] [Accepted: 12/01/2014] [Indexed: 12/19/2022]
Abstract
Premature ovarian failure (POF) is defined as lost ovarian functions before the age of 40. Three possible molecular markers (PLA2G4A, miR-29a, and miR-144) have been identified in our previous study by integrated analysis of mRNA and miRNA expression profiles. The present study aimed to evaluate American ginseng root's protective potential against POF by studying transcriptional and protein variations between American ginseng treatments and controls in rats. 4-Vinylcyclohexene diepoxide (VCD) was administered to rats for 14 days to induce POF. Additionally, American ginseng was administered to POF rats for one month, and PLA2G4A, miR-29a, and miR-144 expressions were measured in rat ovaries by qRT-PCR. PLA2G4A protein expression was examined by Western Blot, and PGE2, LH, FSH, and E2 serum levels were detected by ELISA. PLA2G4A mRNA and protein were downregulated in American ginseng-treated rats, miR-29a and miR-144 levels increased, and PGE2 serum levels decreased, while LH, FSH, and E2 increased compared to POF induction alone. Analysis of transcriptional and protein variations suggested that American ginseng protects the ovary against POF by regulating prostaglandin biosynthesis, ovulation, and preventing ovarian aging. High hormone levels (PGE2, FSH, and LH) were reduced, and E2 secretion approached normal levels, leading to improved POF symptoms and abnormal ovulation.
Collapse
|
21
|
Sayasith K, Sirois J, Lussier JG. Expression and regulation of regulator of G-protein signaling protein-2 (RGS2) in equine and bovine follicles prior to ovulation: molecular characterization of RGS2 transactivation in bovine granulosa cells. Biol Reprod 2014; 91:139. [PMID: 25339105 DOI: 10.1095/biolreprod.114.121186] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The luteinizing hormone preovulatory surge stimulates several signal pathways essential for ovulation, and the regulator of G-protein signaling protein-2 (RGS2) is thought to be involved in this process. The objectives of this study were to characterize the regulation of RGS2 transcripts in equine and bovine follicles prior to ovulation and to determine its transcriptional control in bovine granulosa cells. To assess the regulation of equine RGS2 prior to ovulation, RT-PCR was performed using total RNA extracted from equine follicles collected at various times after human chorionic gonadotropin (hCG) injection. Results showed that RGS2 mRNA levels were very low at 0 h but markedly increased 12-39 h post-hCG (P < 0.05). In the bovine species, results revealed that RGS2 mRNA levels were low in small and dominant follicles and in ovulatory follicles obtained at 0 h, but markedly increased in ovulatory follicles 6-24 h post-hCG (P < 0.05). To study the molecular control of RGS2 expression, primary cultures of bovine granulosa cells were used. Stimulation with forskolin induced an up-regulation of RGS2 mRNA in vitro. Studies using 5'-deletion mutants identified a minimal region containing full-length basal and forskolin-inducible RGS2 promoter activities. Site-directed mutagenesis indicated that these activities were dependent on CRE and ETS1 cis-elements. Electrophoretic mobility shift assays confirmed the involvement of these elements and revealed their interactions with CREB1 and ETS1 proteins. Chromatin immunoprecipitation assays confirmed endogenous interactions of these proteins with the RGS2 promoter in granulosa cells. Forskolin-inducible RGS2 promoter activity and mRNA expression were markedly decreased by PKA and ERK1/2 inhibitors, and treatment with an antagonist of PGR (RU486) and inhibitors of PTGS2 (NS398) and EGFR (PD153035) blocked the forskolin-dependent RGS2 transcript expression, suggesting the importance of RGS2 in ovulation. Collectively, this study reports for the first time the gonadotropin-dependent up-regulation of RGS2 in equine and bovine preovulatory follicles and presents some of the regulatory controls involved in RGS2 gene expression in granulosa cells.
Collapse
Affiliation(s)
- Khampoun Sayasith
- Centre de recherche en reproduction animale and the Département de biomédecine vétérinaire, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | - Jean Sirois
- Centre de recherche en reproduction animale and the Département de biomédecine vétérinaire, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | - Jacques G Lussier
- Centre de recherche en reproduction animale and the Département de biomédecine vétérinaire, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| |
Collapse
|
22
|
Cohen-Fredarow A, Tadmor A, Raz T, Meterani N, Addadi Y, Nevo N, Solomonov I, Sagi I, Mor G, Neeman M, Dekel N. Ovarian dendritic cells act as a double-edged pro-ovulatory and anti-inflammatory sword. Mol Endocrinol 2014; 28:1039-54. [PMID: 24825398 DOI: 10.1210/me.2013-1400] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ovulation and inflammation share common attributes, including immune cell invasion into the ovary. The present study aims at deciphering the role of dendritic cells (DCs) in ovulation and corpus luteum formation. Using a CD11c-EYFP transgenic mouse model, ovarian transplantation experiments, and fluorescence-activated cell sorting analyses, we demonstrate that CD11c-positive, F4/80-negative cells, representing DCs, are recruited to the ovary under gonadotropin regulation. By conditional ablation of these cells in CD11c-DTR transgenic mice, we revealed that they are essential for expansion of the cumulus-oocyte complex, release of the ovum from the ovarian follicle, formation of a functional corpus luteum, and enhanced lymphangiogenesis. These experiments were complemented by allogeneic DC transplantation after conditional ablation of CD11c-positive cells that rescued ovulation. The pro-ovulatory effects of these cells were mediated by up-regulation of ovulation-essential genes. Interestingly, we detected a remarkable anti-inflammatory capacity of ovarian DCs, which seemingly serves to restrict the ovulatory-associated inflammation. In addition to discovering the role of DCs in ovulation, this study implies the extended capabilities of these cells, beyond their classic immunologic role, which is relevant also to other biological systems.
Collapse
Affiliation(s)
- Adva Cohen-Fredarow
- Department of Biological Regulation (A.C.-F., A.T., N.M., Y.A., N.N., I.So., I.Sa., M.N., N.D.), Weizmann Institute of Science, Rehovot 76100, Israel; Koret School of Veterinary Medicine (T.R.), The Hebrew University of Jerusalem, Rehovot 76100, Israel; B-nano Ltd (Y.A.), Rehovot 76326, Israel; and Department of Obstetrics Gynecology and Reproductive Science (G.M.), Reproductive Immunology Unit, Yale University School of Medicine, New Haven, Connecticut 06510
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Park ES, Park J, Franceschi RT, Jo M. The role for runt related transcription factor 2 (RUNX2) as a transcriptional repressor in luteinizing granulosa cells. Mol Cell Endocrinol 2012; 362:165-75. [PMID: 22713854 PMCID: PMC3864655 DOI: 10.1016/j.mce.2012.06.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 05/08/2012] [Accepted: 06/11/2012] [Indexed: 10/28/2022]
Abstract
Transcription factors induced by the LH surge play a vital role in reprogramming the gene expression in periovulatory follicles. The present study investigated the role of RUNX2 transcription factor in regulating the expression of Runx1, Ptgs2, and Tnfaip6 using cultured granulosa cells isolated from PMSG-primed immature rats. hCG or forskolin+PMA induced the transient increase in Runx1, Ptgs2, and Tnfaip6 expression, while the expression of Runx2 continued to increase until 48 h. The knockdown of the agonist-stimulated Runx2 expression increased Runx1, Ptgs2, and Tnfaip6 expression and PGE(2) levels in luteinizing granulosa cells. Conversely, the over-expression of RUNX2 inhibited the expression of these genes and PGE(2) levels. The mutation of RUNX binding motifs in the Runx1 promoter enhanced transcriptional activity of the Runx1 promoter. The knockdown and overexpression of Runx2 increased and decreased Runx1 promoter activity, respectively. ChIP assays revealed the binding of RUNX2 in the Runx1 and Ptgs2 promoters. Together, these novel findings provide support for the role of RUNX2 in down-regulation of Runx1, Ptgs2, and Tnfaip6 during the late ovulatory period to support proper ovulation and/or luteinization.
Collapse
Affiliation(s)
- Eun-Sil Park
- Department of Obstetrics and Gynecology, Chandler Medical Center, University of Kentucky, Lexington, Kentucky 40536-0298, USA
- Department of Molecular and Biomedical Pharmacology, Chandler Medical Center, University of Kentucky, Lexington, Kentucky 40536-0298, USA
| | - Jiyeon Park
- Department of Obstetrics and Gynecology, Chandler Medical Center, University of Kentucky, Lexington, Kentucky 40536-0298, USA
| | - Renny T. Franceschi
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, 1011N University Ave. Ann Arbor, MI 48109-1078, USA
- Department of Biological Chemistry, School of Medicine, University of Michigan, 1011N University Ave. Ann Arbor, MI 48109-1078, USA
| | - Misung Jo
- Department of Obstetrics and Gynecology, Chandler Medical Center, University of Kentucky, Lexington, Kentucky 40536-0298, USA
| |
Collapse
|
24
|
Toda K, Ono M, Yuhki KI, Ushikubi F, Saibara T. 17β-Estradiol is critical for the preovulatory induction of prostaglandin E(2) synthesis in mice. Mol Cell Endocrinol 2012; 362:176-82. [PMID: 22713853 DOI: 10.1016/j.mce.2012.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 06/08/2012] [Accepted: 06/12/2012] [Indexed: 10/28/2022]
Abstract
Aromatase-deficient (ArKO) mice are totally anovulatory due to insufficient estrogen production. However, sequential administrations of high doses of 17β-estradiol (E2) and gonadotropins were found to induce ovulation in these mice. Here, we examined how the ovulatory stimulation for ArKO mice alters the expressions of genes related to prostaglandin (PG) E(2) metabolism and ovarian contents of PGE(2), as PGE(2) is one of the critical mediators of ovulatory induction. The ovulatory stimulation significantly increased mRNA expressions of prostaglandin-endoperoxide synthase 2, PGE(2) receptor type 4 and sulfotransferase family 1E, member 1, in preovulatory ArKO ovaries. In contrast, it suppressed the mRNA expression of 15-hydroxyprostaglandin dehydrogenase. Furthermore, significant elevation in the PGE(2) contents was detected in the preovulatory ovaries of ArKO mice after stimulation with E2 plus ovulatory doses of gonadotropins. Thus, these analyses demonstrate a requirement of E2 for the preovulatory enhancement of PGE(2) synthesis, leading to future success in ovulation.
Collapse
Affiliation(s)
- Katsumi Toda
- Department of Biochemistry, Kochi University, School of Medicine, Nankoku, Kochi 783-8505, Japan.
| | | | | | | | | |
Collapse
|
25
|
Kurusu S, Sapirstein A, Bonventre JV. Group IVA phospholipase A₂ optimizes ovulation and fertilization in rodents through induction of and metabolic coupling with prostaglandin endoperoxide synthase 2. FASEB J 2012; 26:3800-10. [PMID: 22673578 PMCID: PMC3425826 DOI: 10.1096/fj.12-203968] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Accepted: 05/14/2012] [Indexed: 12/12/2022]
Abstract
Female mice lacking group IVA phospholipase A(2) (Pla2g4a(-/-)) have a smaller litter size, which is due, in part, to defective implantation. We examined PLA(2)G4A dependence of the processes of ovulation and fertilization. Following induction of ovulation by equine chorionic gonadotropin (eCG)/human CG (hCG) treatment and mating, ovulation and fertilization rates were reduced significantly in Pla2g4a(-/-) mice as compared to wild-type littermates. Human CG triggered robust ovarian prostaglandin (PG) E(2) production in the preovulatory period that was significantly attenuated in Pla2g4a(-/-) mice. Human CG transiently enhanced ovarian expression of PLA(2)G4A and prostaglandin endoperoxide synthase 2 (PTGS2) in wild-type mice. This PTGS2 induction was decreased in Pla2g4a(-/-) mice and also in immature rats treated with the PLA(2)G4A inhibitor, archidonyl trifluoromethyl ketone. A close spatiotemporal association of PLA(2)G4A with PTGS2 was found in mouse and rat preovulatory follicles examined by immunohistochemistry. Less association was observed with 4 other forms of PLA(2). Our data strongly suggest that PLA(2)G4A amplifies hCG induction of PTGS2 and colocalizes with the induced PTGS2, thus contributing to robust PG production required for optimal ovulation and fertilization in rodents.
Collapse
Affiliation(s)
- Shiro Kurusu
- Department of Medicine and
- Laboratory of Veterinary Physiology, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Adam Sapirstein
- Department of Anesthesia and Critical Care, Massachusetts General Hospital, and
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; and
| | - Joseph V. Bonventre
- Department of Medicine and
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Kyasari O, Valojerdi M, Farrokhi A, Ebrahimi B. Expression of maturation genes and their receptors during in vitro maturation of sheep COCs in the presence and absence of somatic cells of cumulus origin. Theriogenology 2012; 77:12-20. [DOI: 10.1016/j.theriogenology.2011.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 06/21/2011] [Accepted: 07/05/2011] [Indexed: 12/13/2022]
|
27
|
Lee YS, VandeVoort CA, Gaughan JP, Midic U, Obradovic Z, Latham KE. Extensive effects of in vitro oocyte maturation on rhesus monkey cumulus cell transcriptome. Am J Physiol Endocrinol Metab 2011; 301:E196-209. [PMID: 21487073 PMCID: PMC3129840 DOI: 10.1152/ajpendo.00686.2010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The elaboration of a quality oocyte is integrally linked to the correct developmental progression of cumulus cell phenotype. In humans and nonhuman primates, oocyte quality is diminished with in vitro maturation. To determine the changes in gene expression in rhesus monkey cumulus cells (CC) that occur during the final day prior to oocyte maturation and how these changes differ between in vitro (IVM) and in vivo maturation (VVM), we completed a detailed comparison of transcriptomes using the Affymetrix gene array. We observed a large number of genes differing in expression when comparing IVM-CC and VVM-CC directly but a much larger number of differences when comparing the transitions from the prematuration to the post-IVM and post-VVM states. We observed a truncation or delay in the normal pattern of gene regulation but also remarkable compensatory changes in gene expression during IVM. Among the genes affected by IVM are those that contribute to productive cell-cell interactions between cumulus cell and oocyte and between cumulus cells. Numerous genes involved in lipid metabolism are incorrectly regulated during IVM, and the synthesis of sex hormones appears not to be suppressed during IVM. We identified a panel of 24 marker genes, the expression of which should provide the foundation for understanding how IVM can be improved for monitoring IVM conditions and for diagnosing oocyte quality.
Collapse
Affiliation(s)
- Young S Lee
- Fels Institute for Cancer Research & Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | |
Collapse
|
28
|
Nuttinck F, Gall L, Ruffini S, Laffont L, Clement L, Reinaud P, Adenot P, Grimard B, Charpigny G, Marquant-Le Guienne B. PTGS2-Related PGE2 Affects Oocyte MAPK Phosphorylation and Meiosis Progression in Cattle: Late Effects on Early Embryonic Development. Biol Reprod 2011; 84:1248-57. [DOI: 10.1095/biolreprod.110.088211] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
29
|
Atikuzzaman M, Koo OJ, Kang JT, Kwon DK, Park SJ, Kim SJ, Gomez MNL, Oh HJ, Hong SG, Jang G, Lee BC. The 9-cis retinoic acid signaling pathway and its regulation of prostaglandin-endoperoxide synthase 2 during in vitro maturation of pig cumulus cell-oocyte complexes and effects on parthenogenetic embryo production. Biol Reprod 2011; 84:1272-81. [PMID: 21368300 DOI: 10.1095/biolreprod.110.086595] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The addition of 9-cis retinoic acid to the oocyte maturation culture medium has a beneficial effect on in vitro fertilized embryos. However, the mechanism of this activity is not known. Therefore, this study was done to elucidate the effect of 9-cis retinoic acid on parthenogenetic embryo production and its signaling pathway and molecular function during in vitro maturation of porcine cumulus cell-oocyte complexes (COCs). Concentrations of 0, 5, 50, and 500 nM 9-cis retinoic acid were added to the in vitro maturation medium, and the embryos were assessed after parthenogenetic activation. Cumulus cells and oocytes from the in vitro matured COCs were separated and subjected to RT-PCR and real-time RT-PCR for detecting retinoic acid receptors and measuring expression of prostaglandin-endoperoxide synthase1 and 2. The addition of 5 nM 9-cis retinoic acid to the maturation medium was beneficial for parthenogenetic embryo production. The effect of 9-cis retinoic acid was exerted directly through the oocytes via the retinoic acid receptor alpha and retinoid X receptor gamma signaling pathways and indirectly through the cumulus cells by the retinoic acid receptor beta and gamma and retinoid X receptor alpha and beta signaling pathways. The addition of 5 nM 9-cis retinoic acid-stimulated cumulus cells reaches full expansion by suppressing their excessive expression of prostaglandin-endoperoxide synthase 2. This study shows that 9-cis retinoic acid can exert its beneficial effect on parthenogenetic embryo production in pigs by multidimensional pathways affecting oocyte maturation.
Collapse
Affiliation(s)
- Mohammad Atikuzzaman
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
The cumulus cell gene expression profile of oocytes with different nuclear maturity and potential for blastocyst formation. J Assist Reprod Genet 2010; 28:31-40. [PMID: 20859762 DOI: 10.1007/s10815-010-9481-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 09/05/2010] [Indexed: 10/19/2022] Open
Abstract
PURPOSE Gene expression in human ART cumulus cell (CC) has been related to oocyte maturity and competence but requires further validation. Expression dynamics were investigated in CC of oocytes at different maturational stages and with different developmental competence in a standard in vivo mouse superovulation model. METHODS Quantitative PCR analysis of Has2, Vcan, Sdc4, Alcam, Grem1, Ptgs1 and Ptgs2 in CC collected at regular time intervals from 0 to 24 h post hCG injection. RESULTS Three expression patterns were observed each with strong regulation (4-230× differences). Immediately prior to ovulation CC of GVBD oocytes have 5× less Sdc4 and Ptgs1 and 5× more Ptgs2 when compared to the CC of freshly ovulated PB oocytes. When compared to the latter, the post-ovulatory aged PB oocytes had a 2× reduced blastocyst forming capacity and their CC expressed 2× more Sdc4 and 6× less Alcam. CONCLUSIONS Morphologically identical cumulus oocyte complexes with different developmental competence can be differentiated by CC gene expression.
Collapse
|
31
|
Boyer A, Lapointe É, Zheng X, Cowan RG, Li H, Quirk SM, DeMayo FJ, Richards JS, Boerboom D. WNT4 is required for normal ovarian follicle development and female fertility. FASEB J 2010; 24:3010-25. [PMID: 20371632 PMCID: PMC2909279 DOI: 10.1096/fj.09-145789] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Accepted: 03/11/2010] [Indexed: 12/21/2022]
Abstract
To study the role of WNT4 in the postnatal ovary, a mouse strain bearing a floxed Wnt4 allele was created and mated to the Amhr2(tm3(cre)Bhr) strain to target deletion of Wnt4 to granulosa cells. Wnt4(flox/-);Amhr2(tm3(cre)Bhr/+) mice had reduced ovary weights and produced smaller litters (P<0.05). Serial follicle counting demonstrated that Wnt4(flox/-);Amhr2(tm3(cre)Bhr/+) mice were born with a normal ovarian reserve and maintained normal numbers of small follicles until puberty but had only 25.2% of the normal number of healthy antral follicles. Some Wnt4(flox/-);Amhr2(tm3(cre)Bhr/+) mice had no antral follicles or corpora lutea and underwent premature follicle depletion. RT-PCR analyses of Wnt4(flox/-);Amhr2(tm3(cre)Bhr/+) granulosa cells and cultured granulosa cells that overexpress WNT4 demonstrated that WNT4 regulates the expression of Star, Cyp11a1, and Cyp19, steroidogenic genes previously identified as downstream targets of the WNT signaling effector CTNNB1. Decreased serum progesterone levels were found in immature, gonadotropin-treated Wnt4(flox/-);Amhr2(tm3(cre)Bhr/+) mice (P<0.05). WNT4- and CTNNB1-overexpressing cultured granulosa cells were analyzed by microarray for alterations in gene expression, which showed that WNT4 regulates additional genes involved in late follicle development via the WNT/CTNNB1 signaling pathway. Together, these data indicate that WNT4 is required for normal antral follicle development and may act by regulating granulosa cell functions including steroidogenesis.
Collapse
Affiliation(s)
- Alexandre Boyer
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; and
| | - Évelyne Lapointe
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | - Xiaofeng Zheng
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | - Robert G. Cowan
- Department of Animal Science, Cornell University, Ithaca, New York, USA
| | - Huaiguang Li
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; and
| | - Susan M. Quirk
- Department of Animal Science, Cornell University, Ithaca, New York, USA
| | - Francesco J. DeMayo
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; and
| | - JoAnne S. Richards
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; and
| | - Derek Boerboom
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; and
| |
Collapse
|
32
|
Kim SY, Weiss J, Tong M, Laronda MM, Lee EJ, Jameson JL. Foxl2, a forkhead transcription factor, modulates nonclassical activity of the estrogen receptor-alpha. Endocrinology 2009; 150:5085-93. [PMID: 19797124 PMCID: PMC2775987 DOI: 10.1210/en.2009-0313] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Foxl2 is a forkhead transcription factor required for ovary development and ovarian follicle maturation. In this report, we identified and characterized a functional relationship between Foxl2 expression and estrogen receptor (ER)-alpha signaling. We show that Foxl2 has no effect on classical ERalpha-mediated transcription, which occurs through canonical estrogen response elements. However, Foxl2 suppresses ERalpha signaling through nonclassical tethered transcriptional pathways. Specifically, the selective ER modulator tamoxifen stimulates activator protein-1 (AP1)-dependent transcription via the ERalpha, and this enhancement is blocked by Foxl2. Two lines of evidence suggest that Foxl2 suppression is mediated by physical interactions with ERalpha rather than direct action at AP1 binding sites. First, ERalpha is coimmunoprecipitated with Foxl2. Second, activation of a upstream activating sequence (UAS) reporter by Gal4-cJun in the presence of ERalpha and tamoxifen was blocked by Foxl2, demonstrating suppression in the absence of an AP1 site. Cyclooxygenase-2 (COX2), which is required for ovulation, was identified through expression profiling as a candidate physiological target for nonclassical ERalpha signaling and thus modulation by ERalpha/Foxl2 interactions. This possibility was confirmed by two sets of experiments. COX2 protein levels were induced by ERalpha in the presence of tamoxifen, and protein expression was suppressed by Foxl2. In addition, ERalpha stimulation of the COX2 promoter was repressed by Foxl2. We conclude that ERalpha and Foxl2 interact and that Foxl2 selectively suppresses ERalpha-mediated transcription of AP1-regulated genes. These data provide a potential point of convergence for ERalpha and Foxl2 to regulate ovarian development and function.
Collapse
Affiliation(s)
- So-Youn Kim
- Morton 4-656, 303 East Chicago Avenue, Chicago, Illinois 60611, USA
| | | | | | | | | | | |
Collapse
|
33
|
Liu J, Park ES, Jo M. Runt-related transcription factor 1 regulates luteinized hormone-induced prostaglandin-endoperoxide synthase 2 expression in rat periovulatory granulosa cells. Endocrinology 2009; 150:3291-300. [PMID: 19342459 PMCID: PMC2703554 DOI: 10.1210/en.2008-1527] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Runt-related transcription factor 1 (RUNX1), a transcription factor, is transiently induced by the LH surge and regulates gene expression in periovulatory granulosa cells. Potential binding sites for RUNX are present in the 5'-flanking region of the Ptgs2 (prostaglandin-endoperoxide synthase 2) gene. Periovulatory Ptgs2 expression is essential for ovulation. In the present study, we investigated the role of RUNX1 in mediating the LH-induced expression of Ptgs2 in periovulatory granulosa cells. We first determined whether the suppression of Runx1 expression or activity affects Ptgs2 expression using cultured preovulatory granulosa cells isolated from immature rat ovaries primed with pregnant mare serum gonadotropin for 48 h. Knockdown of human chorionic gonadotropin-induced Runx1 expression by small interfering RNA or inhibition of endogenous RUNX activities by dominant-negative RUNX decreased human chorionic gonadotropin or agonist-stimulated Ptgs2 expression and transcriptional activity of Ptgs2 promoter reporter constructs. Results from chromatin immunoprecipitation assays revealed in vivo binding of endogenous RUNX1 to the Ptgs2 promoter region in rat periovulatory granulosa cells. Direct binding of RUNX1 to two RUNX-binding motifs in the Ptgs2 promoter region was confirmed by EMSA. The mutation of these two binding motifs resulted in decreased transcriptional activity of Ptgs2 promoter reporter constructs in preovulatory granulosa cells. Taken together, these findings provide experimental evidence that the LH-dependent induction of Ptgs2 expression results, in part, from RUNX1-mediated transactivation of the Ptgs2 promoter. The results of the present study assign potential significance for LH-induced RUNX1 in the ovulatory process via regulating Ptgs2 gene expression.
Collapse
Affiliation(s)
- Jing Liu
- Department of Obstetrics and Gynecology, Chandler Medical Center, 800 Rose Street, University of Kentucky, Lexington, Kentucky 40536-0298, USA
| | | | | |
Collapse
|
34
|
Caillaud M, Gérard N. In vivo and in vitro effects of interleukin-1beta on equine oocyte maturation and on steroidogenesis and prostaglandin synthesis in granulosa and cumulus cells. Reprod Fertil Dev 2009; 21:265-73. [PMID: 19210917 DOI: 10.1071/rd08046] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Accepted: 09/08/2008] [Indexed: 11/23/2022] Open
Abstract
We analysed the effect of interleukin-1 on oocyte maturation and on steroid and prostaglandin production by equine granulosa and cumulus cells. In Experiment 1, interleukin-1beta (IL-1beta) was injected into the growing dominant follicle, which was punctured 38 h later. Follicular fluid was assayed for steroids and prostaglandin-F2alpha (PGF2alpha). Granulosa cells were analysed for 3beta-hydroxysteroid dehydrogenase (3beta-HSD), progesterone receptor (PR), cyclooxygenase 1 and 2 (Cox 1 and Cox 2) and steroidogenic acute regulatory protein (StAR) mRNAs. In Experiment 2, cumulus-oocyte complexes (COCs) were collected from slaughterhouse ovaries and cultured in different media: control group (TCM199 + BSA); Group 2 (+ IL-1beta); Group 3 (+ EGF); Group 4 (+ EGF + IL-1beta); and Group 5 (+ EGF + IL-1beta + IL-1RA). Cumulus cells were analysed for 3beta-HSD, PR, Cox 1, Cox 2 and StAR mRNAs. After injections of crude equine gonadotropin (CEG; LH effect) or IL-1beta, progesterone and PGF2alpha levels increased, whereas 17beta-oestradiol decreased. EGF induced an increase in the rate of in vitro maturation (P < 0.05), whereas IL-1beta had a limited effect. IL-1beta significantly decreased the rate of EGF-induced oocyte maturation (P < 0.05). Cox 2 mRNA level increases in granulosa cells after CEG injection (P = 0.07). In cumulus cells, StAR and PR mRNAs were lower in Group 2 and 3beta-HSD mRNA was higher in Groups 4 and 5. These data confirm that IL-1 is involved in equine oocyte in vitro maturation. We demonstrated in vivo that IL-1beta has an effect on steroids and PGF2alpha secretion in the preovulatory follicle.
Collapse
Affiliation(s)
- Maud Caillaud
- INRA, UMR 85 Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France
| | | |
Collapse
|
35
|
Su YQ, Sugiura K, Eppig JJ. Mouse oocyte control of granulosa cell development and function: paracrine regulation of cumulus cell metabolism. Semin Reprod Med 2009; 27:32-42. [PMID: 19197803 PMCID: PMC2742468 DOI: 10.1055/s-0028-1108008] [Citation(s) in RCA: 260] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Bidirectional communication between oocytes and the companion granulosa cells is essential for the development and functions of both compartments. Oocytes are deficient in their ability to transport certain amino acids and in carrying out glycolysis and cholesterol biosynthesis. Cumulus cells must provide them with the specific amino acids and the products in these metabolic pathways. Oocytes control metabolic activities in cumulus cells by promoting the expression of genes in cumulus cells encoding specific amino acid transporters and enzymes essential for the oocyte-deficient metabolic processes. Hence oocytes outsource metabolic functions to cumulus cells to compensate for oocyte metabolic deficiencies. Oocyte control of granulosa cell metabolism may also participate in regulating the rate of follicular development in coordination with endocrine, paracrine, and autocrine signals. Oocytes influence granulosa cell development mainly by secretion of paracrine factors, although juxtacrine signals probably also participate. Key oocyte-derived paracrine factors include growth differentiation factor 9, bone morphogenetic protein 15, and fibroblast growth factor 8B.
Collapse
Affiliation(s)
- You-Qiang Su
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA.
| | | | | |
Collapse
|
36
|
Gilchrist RB, Lane M, Thompson JG. Oocyte-secreted factors: regulators of cumulus cell function and oocyte quality. Hum Reprod Update 2008; 14:159-77. [PMID: 18175787 DOI: 10.1093/humupd/dmm040] [Citation(s) in RCA: 670] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Oocyte quality is a key limiting factor in female fertility, yet we have a poor understanding of what constitutes oocyte quality or the mechanisms governing it. The ovarian follicular microenvironment and maternal signals, mediated primarily through granulosa cells (GCs) and cumulus cells (CCs), are responsible for nurturing oocyte growth, development and the gradual acquisition of oocyte developmental competence. However, oocyte-GC/CC communication is bidirectional with the oocyte secreting potent growth factors that act locally to direct the differentiation and function of CCs. Two important oocyte-secreted factors (OSFs) are growth-differentiation factor 9 and bone morphogenetic protein 15, which activate signaling pathways in CCs to regulate key genes and cellular processes required for CC differentiation and for CCs to maintain their distinctive phenotype. Hence, oocytes appear to tightly control their neighboring somatic cells, directing them to perform functions required for appropriate development of the oocyte. This oocyte-CC regulatory loop and the capacity of oocytes to regulate their own microenvironment by OSFs may constitute important components of oocyte quality. In support of this notion, it has recently been demonstrated that supplementing oocyte in vitro maturation (IVM) media with exogenous OSFs improves oocyte developmental potential, as evidenced by enhanced pre- and post-implantation embryo development. This new perspective on oocyte-CC interactions is improving our knowledge of the processes regulating oocyte quality, which is likely to have a number of applications, including improving the efficiency of clinical IVM and thereby providing new options for the treatment of infertility.
Collapse
Affiliation(s)
- Robert B Gilchrist
- Research Centre for Reproductive Health, School of Paediatrics and Reproductive Health, Discipline of Obstetrics and Gynaecology, Medical School, University of Adelaide, Adelaide 5005, Australia.
| | | | | |
Collapse
|
37
|
Peroxisome proliferator-activated receptor gamma is a target of progesterone regulation in the preovulatory follicles and controls ovulation in mice. Mol Cell Biol 2008; 28:1770-82. [PMID: 18172011 DOI: 10.1128/mcb.01556-07] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The progesterone receptor (PR) plays a critical role during ovulation. Mice lacking the PR gene are anovulatory due to a failure in the rupture of the preovulatory follicles. The pathways that operate downstream of PR to control ovulation are poorly understood. Using gene expression profiling, we identified peroxisome proliferator-activated receptor gamma (PPARgamma) as a target of regulation by PR in the granulosa cells of the preovulatory follicles during the ovulatory process. To investigate the function of PPARgamma during ovulation, we created a conditional knockout mouse in which this gene was deleted via Cre-Lox-mediated excision in granulosa cells. When these mutant mice were subjected to gonadotropin-induced superovulation, the preovulatory follicles failed to rupture and the number of eggs released from the mutant ovaries declined drastically. Gene expression analysis identified endothelin-2, interleukin-6, and cyclic GMP-dependent protein kinase II as novel targets of regulation by PPARgamma in the ovary. Our studies also suggested that cycloxygenase 2-derived metabolites of long-chain fatty acids function as endogenous activating ligands of PPARgamma in the preovulatory follicles. Collectively, these studies revealed that PPARgamma is a key mediator of the biological actions of PR in the granulosa cells and activation of its downstream pathways critically controls ovulation.
Collapse
|
38
|
Su YQ, Sugiura K, Wigglesworth K, O'Brien MJ, Affourtit JP, Pangas SA, Matzuk MM, Eppig JJ. Oocyte regulation of metabolic cooperativity between mouse cumulus cells and oocytes: BMP15 and GDF9 control cholesterol biosynthesis in cumulus cells. Development 2007; 135:111-21. [PMID: 18045843 DOI: 10.1242/dev.009068] [Citation(s) in RCA: 269] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Oocyte-derived bone morphogenetic protein 15 (BMP15) and growth differentiation factor 9 (GDF9) are key regulators of follicular development. Here we show that these factors control cumulus cell metabolism, particularly glycolysis and cholesterol biosynthesis before the preovulatory surge of luteinizing hormone. Transcripts encoding enzymes for cholesterol biosynthesis were downregulated in both Bmp15(-/-) and Bmp15(-/-) Gdf9(+/-) double mutant cumulus cells, and in wild-type cumulus cells after removal of oocytes from cumulus-cell-oocyte complexes. Similarly, cholesterol synthesized de novo was reduced in these cumulus cells. This indicates that oocytes regulate cumulus cell cholesterol biosynthesis by promoting the expression of relevant transcripts. Furthermore, in wild-type mice, Mvk, Pmvk, Fdps, Sqle, Cyp51, Sc4mol and Ebp, which encode enzymes required for cholesterol synthesis, were highly expressed in cumulus cells compared with oocytes; and oocytes, in the absence of the surrounding cumulus cells, synthesized barely detectable levels of cholesterol. Furthermore, coincident with reduced cholesterol synthesis in double mutant cumulus cells, lower levels were also detected in cumulus-cell-enclosed double mutant oocytes compared with wild-type oocytes. Levels of cholesterol synthesis in double mutant cumulus cells and oocytes were partially restored by co-culturing with wild-type oocytes. Together, these results indicate that mouse oocytes are deficient in synthesizing cholesterol and require cumulus cells to provide products of the cholesterol biosynthetic pathway. Therefore, oocyte-derived paracrine factors, particularly, BMP15 and GDF9, promote cholesterol biosynthesis in cumulus cells, probably as compensation for oocyte deficiencies in cholesterol production.
Collapse
Affiliation(s)
- You-Qiang Su
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Russell DL, Robker RL. Molecular mechanisms of ovulation: co-ordination through the cumulus complex. Hum Reprod Update 2007; 13:289-312. [PMID: 17242016 DOI: 10.1093/humupd/dml062] [Citation(s) in RCA: 275] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Successful ovulation requires that developmentally competent oocytes are released with appropriate timing from the ovarian follicle. Somatic cells of the follicle sense the ovulatory stimulus and guide resumption of meiosis and release of the oocyte, as well as structural remodelling and luteinization of the follicle. Complex intercellular communication co-ordinates critical stages of oocyte maturation and links this process with release from the follicle. To achieve these outcomes, ovulation is controlled through multiple inputs, including endocrine hormones, immune and metabolic signals, as well as intrafollicular paracrine factors from the theca, mural and cumulus granulosa cells and the oocyte itself. This review focuses on the recent advances in understanding of molecular mechanisms that commence after the gonadotrophin surge and culminate with release of the oocyte. These mechanisms include intracellular signalling, gene regulation and remodelling of tissue structure in each of the distinct ovarian compartments. Most critical ovulatory mediators exert effects through the cumulus cell complex that surrounds and connects with the oocyte. The convergence of ovulatory signals through the cumulus complex co-ordinates the key mechanistic processes that mediate and control oocyte maturation and ovulation.
Collapse
Affiliation(s)
- Darryl L Russell
- Research Centre for Reproductive Health, School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, South Australia, Australia.
| | | |
Collapse
|
40
|
Shimada M, Hernandez-Gonzalez I, Gonzalez-Robanya I, Richards JS. Induced Expression of Pattern Recognition Receptors in Cumulus Oocyte Complexes: Novel Evidence for Innate Immune-Like Functions during Ovulation. Mol Endocrinol 2006; 20:3228-39. [PMID: 16931571 DOI: 10.1210/me.2006-0194] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Ovulation is the complex, inflammatory-like process by which the cumulus oocyte complex (COC) is released from a mature, preovulatory follicle through a rupture site at the ovarian surface and requires expression of genes that generate and stabilize the expanded extracellular COC matrix. Gene profiling analyses of COCs at selected time intervals during ovulation revealed that many genes associated with immune related surveillance functions were also induced in cumulus cells. Specifically, cell surface signaling molecules known as pattern recognition receptors that act as sensors of the external environment important for the innate immune system to detect self from nonself or altered self are induced and/or expressed in cumulus cells as well as granulosa cells. These include the complement factor q1, CD14, and the Toll-like receptors (TLRs) 4, 8, and 9 as well as mediators of TLR activation, myeloid differentiation primary response gene 88 and interferon regulatory factor 3. COCs exposed to bacterial lipopolysaccharide exhibit enhanced phosphorylation of p38MAPK, ERK1/2 and nuclear factor-kappaB and increased expression of Il6 and Tnfa target genes, documenting that the TLR pathway is functional. Cumulus cells and granulosa cells also express the scavenger receptors CD36 and scavenger receptor type B1 and exhibited phagocytic uptake of fluorescently tagged bacterial particles. Collectively, these results provide novel evidence that cumulus cells as well as granulosa cells express innate immune related genes that may play critical roles in surveillance and cell survival during the ovulation process.
Collapse
Affiliation(s)
- Masayuki Shimada
- Department of Molecular Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
41
|
Takahashi T, Morrow JD, Wang H, Dey SK. Cyclooxygenase-2-derived Prostaglandin E2 Directs Oocyte Maturation by Differentially Influencing Multiple Signaling Pathways. J Biol Chem 2006; 281:37117-29. [PMID: 17023426 DOI: 10.1074/jbc.m608202200] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The process of oocyte maturation, which impacts ovulation and fertilization, is complex and requires an integration of the endocrine, paracrine, juxtacrine, and autocrine signaling pathways. This process involves an intimate interaction between the oocyte and encircling cumulus cells within a follicle, a unique venue for somatic and germ cell communication. Cumulus cell expansion and resumption of meiosis with germinal vesicle breakdown are major events in oocyte maturation. Cyclooxygenase-2 (COX-2)-derived prostaglandin E(2) (PGE(2)) is a known critical mediator of oocyte maturation, but the diverse function of this lipid mediator in oocyte maturation, ovulation, and fertilization has not been fully appreciated. We show here that gonadotropins in coordination with PGE(2) signaling via its cell surface G-protein-coupled EP2 and EP4 receptor subtypes direct cumulus cell expansion and survival and oocyte meiotic maturation by differentially impacting cAMP-dependent protein kinase, MAPK, NF-kappaB, and phosphatidylinositol 3-kinase/Akt pathways. This study is unique in the sense that it provides evidence for new site- and event-specific involvement of these signaling pathways under the influence of COX-2-derived PGE(2) during the critical stages of this somatic-germ cell interaction, an absolute requirement for oocyte maturation.
Collapse
Affiliation(s)
- Toshifumi Takahashi
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | | | |
Collapse
|
42
|
Xing Y, Gosden R, Lasko P, Clarke H. Murine homologues of the Drosophila gustavus gene are expressed in ovarian granulosa cells. Reproduction 2006; 131:905-15. [PMID: 16672355 PMCID: PMC5123870 DOI: 10.1530/rep.1.01046] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mammalian homologues of genes that control oogenesis in other organisms may play similar roles in mammalian ovarian development. In Drosophila melanogaster, GUSTAVUS (GUS) protein physically interacts with and is necessary for the proper posterior localization of VASA protein, and thus is required for specification of germ cells. We identified two mouse genes, SSB-1 and SSB-4 (SPRY domain SOCS box protein), whose protein products share 75% identity and are each approximately 70% identical to Drosophila GUS. Both SSB-1 and SSB-4 mRNA were detectable in mouse ovaries by Northern blotting of total and poly(A) + RNA, but were expressed in few other tissues. SSB-1 was detectable in testes, although the 3'-untranslated region of the mRNA was considerably shorter than the ovarian mRNA. In situ hybridization and RT-PCR analysis of ovaries revealed that both genes were expressed in granulosa cells at all stages of follicular development. In contrast, expression was barely detectable in in oocytes. Immunoblotting analysis revealed that SSB-1 protein was present in follicles at different stages of growth, and immunocytochemistry confirmed that SSB-1 and SSB-4 were detectable in granulosa cells of primary and subsequent stage follicles and that they were present in both mural and cumulus granulosa cells of antral follicles. These results establish that GUS-related proteins, which in Drosophila are restricted to the germ cells, are in the mouse instead expressed in the granulosa cells and are present throughout folliculogenesis. Based on their tissue-restricted pattern of expression and apparent abundance in granulosa cells, we propose that SSB-1 and SSB-4 play key roles in regulating granulosa cell physiology.
Collapse
Affiliation(s)
- Yan Xing
- Department of Obstetrics and Gynecology, McGill University, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Roger Gosden
- Center for Reproductive Medicine and Infertility, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Paul Lasko
- Department of Biology, McGill University, Montreal, Quebec, Canada
| | - Hugh Clarke
- Department of Obstetrics and Gynecology, McGill University, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Biology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
43
|
Diaz FJ, O'Brien MJ, Wigglesworth K, Eppig JJ. The preantral granulosa cell to cumulus cell transition in the mouse ovary: Development of competence to undergo expansion. Dev Biol 2006; 299:91-104. [PMID: 16908014 DOI: 10.1016/j.ydbio.2006.07.012] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2006] [Revised: 06/29/2006] [Accepted: 07/11/2006] [Indexed: 11/20/2022]
Abstract
The transition of preantral to antral follicles is one of the major steps in follicular development, yet little is known about the molecular and functional changes that occur as preantral granulosa cells differentiate into cumulus cells. The cumulus oophorus of large antral follicles undergoes expansion in response to the preovulatory surge of gonadotropins, but preantral granulosa cells do not. The objective of this project was to determine the molecular mechanisms underlying this differential response. Cumulus expansion in vitro requires secretion of cumulus-expansion enabling factors (CEEFs) by the oocyte and stimulation by a ligand, epidermal growth factor (EGF) or follicle-stimulating hormone (FSH). This combined stimulation results in activation of MAPKs (MAPK3/1 (formerly ERK1/2) and MAPK14 (formerly p38)) and increased Has2, Ptgs2, Tnfaip6 and Ptx3 mRNA levels, all of which are required for cumulus expansion. Only fully-grown oocytes from antral follicles were competent to enable expansion and increases in expansion-related transcripts in cumulus cells, whereas growing oocytes of preantral follicles did not. To assess the competence of preantral granulosa cells to generate responses associated with expansion, they were treated with FSH or EGF and co-cultured with fully-grown oocytes secreting CEEFs. MAPKs were activated by EGF in preantral granulosa cells to essentially the same levels as in cumulus cells. Preantral granulosa cells treated with EGF, but not those treated with FSH increased Has2, Ptgs2 and Ptx3 mRNAs to 17-96% of the levels observed in cumulus cells. In contrast, the level of Tnfaip6 mRNA was minimally stimulated in preantral granulosa cells. Therefore, preantral granulosa cells do not undergo expansion for two fundamental reasons. First, the growing oocytes of preantral follicles do not secrete active CEEFs. Second, activation of MAPKs alone in preantral granulosa cells, even in the presence of CEEFs, is not sufficient to increase the expression of essential transcripts, particularly Tnfaip6 mRNA. Thus, preantral granulosa cells differ from cumulus cells in CEEF-dependent processes downstream of the activation of MAPKs.
Collapse
Affiliation(s)
- F J Diaz
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | | | | | |
Collapse
|
44
|
Localization of cystathionine β synthase in mice ovaries and its expression profile during follicular development. Chin Med J (Engl) 2006. [PMID: 17134586 DOI: 10.1097/00029330-200611020-00006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
45
|
Su YQ, Sugiura K, Woo Y, Wigglesworth K, Kamdar S, Affourtit J, Eppig JJ. Selective degradation of transcripts during meiotic maturation of mouse oocytes. Dev Biol 2006; 302:104-17. [PMID: 17022963 PMCID: PMC1847322 DOI: 10.1016/j.ydbio.2006.09.008] [Citation(s) in RCA: 251] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2006] [Revised: 08/31/2006] [Accepted: 09/01/2006] [Indexed: 11/28/2022]
Abstract
There is massive destruction of transcripts during the maturation of mouse oocytes. The objective of this project was to identify and characterize the transcripts that are degraded versus those that are stable during the transcriptionally silent germinal vesicle (GV)-stage to metaphase II (MII)-stage transition using a microarray approach. A system for oocyte transcript amplification using both internal and 3'-poly(A) priming was utilized to minimize the impact of complex variations in transcript polyadenylation prevalent during this transition. Transcripts were identified and quantified using the Affymetrix Mouse Genome 430 v2.0 GeneChip. The significantly changed and stable transcripts were analyzed using Ingenuity Pathways Analysis and GenMAPP/MAPPFinder to characterize the biological themes underlying global changes in oocyte transcripts during maturation. It was concluded that the destruction of transcripts during the GV to MII transition is a selective rather than promiscuous process in mouse oocytes. In general, transcripts involved in processes that are associated with meiotic arrest at the GV-stage and the progression of oocyte maturation, such as oxidative phosphorylation, energy production, and protein synthesis and metabolism, were dramatically degraded. In contrast, transcripts encoding participants in signaling pathways essential for maintaining the unique characteristics of the MII-arrested oocyte, such as those involved in protein kinase pathways, were the most prominent among the stable transcripts.
Collapse
Affiliation(s)
- You-Qiang Su
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609 United States
| | - Koji Sugiura
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609 United States
| | - Yong Woo
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609 United States
- Functional Genomics Program, The University of Maine, Orono, ME 04469, United States
| | - Karen Wigglesworth
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609 United States
| | - Sonya Kamdar
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609 United States
| | - Jason Affourtit
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609 United States
| | - John J. Eppig
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609 United States
- *Corresponding author: Fax: 1 207 288 6073, E-mail address:
| |
Collapse
|
46
|
Shimada M, Hernandez-Gonzalez I, Gonzalez-Robayna I, Richards JS. Paracrine and autocrine regulation of epidermal growth factor-like factors in cumulus oocyte complexes and granulosa cells: key roles for prostaglandin synthase 2 and progesterone receptor. Mol Endocrinol 2006; 20:1352-65. [PMID: 16543407 DOI: 10.1210/me.2005-0504] [Citation(s) in RCA: 329] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The molecular bridges that link the LH surge with functional changes in cumulus cells that possess few LH receptors are being unraveled. Herein we document that epidermal growth factor (EGF)-like factors amphiregulin (Areg), epiregulin (Ereg), and betacellulin (Btc) are induced in cumulus oocyte complexes (COCs) by autocrine and paracrine mechanisms that involve the actions of prostaglandins (PGs) and progesterone receptor (PGR). Areg and Ereg mRNA and protein levels were reduced significantly in COCs and ovaries collected from prostaglandin synthase 2 (Ptgs2) null mice and Pgr null (PRKO) mice at 4 h and 8 h after human chorionic gonadotropin, respectively. In cultured COCs, FSH/forskolin induced Areg mRNA within 0.5 h that peaked at 4 h, a process blocked by inhibitors of p38MAPK (SB203580), MAPK kinase (MEK) 1 (PD98059), and PTGS2 (NS398) but not protein kinase A (PKA) (KT5720). Conversely, AREG but not FSH induced Ptsg2 mRNA at 0.5 h with peak expression of Ptgs2 and Areg mRNAs at 4 h, processes blocked by the EGF receptor tyrosine kinase inhibitor AG1478 (AG), PD98059, and NS398. PGE2 reversed the inhibitory effects of AG on AREG-induced expression of Areg but not Ptgs2, placing Ptgs2 downstream of EGF-R signaling. Phorbol 12-myristate 13-acetate (PMA) and adenovirally expressed PGRA synergistically induced Areg mRNA in granulosa cells. In COCs, AREG not only induced genes that impact matrix formation but also genes involved in steroidogenesis (StAR, Cyp11a1) and immune cell-like functions (Pdcd1, Runx1, Cd52). Collectively, FSH-mediated induction of Areg mRNA via p38MAPK precedes AREG induction of Ptgs2 mRNA via ERK1/2. PGs acting via PTGER2 in cumulus cells provide a secondary, autocrine pathway to regulate expression of Areg in COCs showing critical functional links between G protein-coupled receptor and growth factor receptor pathways in ovulating follicles.
Collapse
Affiliation(s)
- Masayuki Shimada
- Department of Molecular Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
47
|
Motola S, Cao X, Ashkenazi H, Popliker M, Tsafriri A. GnRH actions on rat preovulatory follicles are mediated by paracrine EGF-like factors. Mol Reprod Dev 2006; 73:1271-6. [PMID: 16865724 DOI: 10.1002/mrd.20556] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Gonadotropin releasing hormone (GnRH) has been shown to mimic the actions of LH/hCG on oocyte maturation and ovulation. Recent studies demonstrated that induction of ovulation by LH/hCG is mediated, at least in part, by transactivation of epidermal growth factor receptors (EGFR) by autocrine/paracrine EGF-like factors activated by metalloproteases. Here we have examined whether the action of GnRH on the preovulatory follicles is exerted through similar mechanisms involving activation of EGFR. The EGFR kinase inhibitor, AG1478, inhibited GnRH-induced oocyte maturation in explanted follicles in vitro. Its inactive analog, AG43, did not affect GnRH-stimulated resumption of meiosis. GnRH, like LH, stimulated transient follicular expression of EGF-like agents, as well as rat cycloxygenase-2 (rCOX-2), rat hyaluronan synthase-2 (rHAS-2), and rat tumor necrosis factor-alpha-stimulated gene 6 (rTSG-6) mRNAs, known ovulatory enzymes. Likewise, GnRH stimulated follicular progesterone synthesis. Conversely AG1478 inhibited all these actions of GnRH. Furthermore, Galardin, a broad-spectrum metalloprotease inhibitor, blocked GnRH-induced oocyte maturation and follicular progesterone synthesis. In conclusion, we have demonstrated that follicular EGF-like factors mediate also the GnRH-stimulation of ovulatory changes, like these of LH/hCG.
Collapse
Affiliation(s)
- Shmulik Motola
- The Bernhard Zondek Hormone Research Laboratory, Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | |
Collapse
|
48
|
Pakarainen T, Zhang FP, Nurmi L, Poutanen M, Huhtaniemi I. Knockout of Luteinizing Hormone Receptor Abolishes the Effects of Follicle-Stimulating Hormone on Preovulatory Maturation and Ovulation of Mouse Graafian Follicles. Mol Endocrinol 2005; 19:2591-602. [PMID: 15941853 DOI: 10.1210/me.2005-0075] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
It is considered a dogma that a secretory peak of LH is indispensable as the trigger of ovulation. However, earlier studies on hypophysectomized rodents have shown that stimulation with recombinant FSH, devoid of any LH activity, is able to boost the final stages of follicular maturation and trigger ovulation. As the expression of ovarian LH receptors (LHRs) still persists after hypophysectomy, such studies cannot totally exclude the possibility that LHR activation is involved in the apparently pure FSH effects. To revisit this question, we analyzed in LHR knockout (LuRKO) mice the progression of folliculogenesis and induction of ovulation by human chorionic gonadotropin and human recombinant FSH treatments. The results provide clear evidence that follicular development and ovulation could not be induced by high doses of FSH in the absence of LHR expression. Ovarian histology and oocyte analyses indicated that follicular maturation did not advance in LuRKO mice beyond the antral follicle stage. Neither were ovulations detected in LuRKO ovaries after any of the gonadotropin treatments. The ovarian resistance to FSH treatment in the absence of LHR was confirmed by real-time RT-PCR and immunohistochemical analyses of a number of gonadotropin-dependent genes, which only responded to the treatments in wild-type control mice. Negative findings were not altered by estradiol priming preceding the gonadotropin stimulations. Hence, the present study shows that, in addition to ovulation, the expression of LHR is essential for follicular maturation in the progression from antral to preovulatory stage.
Collapse
Affiliation(s)
- Tomi Pakarainen
- Department of Physiology, University of Turku, Fin-20500 Turku, Finland
| | | | | | | | | |
Collapse
|
49
|
Irving-Rodgers HF, Rodgers RJ. Extracellular matrix in ovarian follicular development and disease. Cell Tissue Res 2005; 322:89-98. [PMID: 16158327 DOI: 10.1007/s00441-005-0042-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Accepted: 07/01/2005] [Indexed: 11/30/2022]
Abstract
The ovarian follicle contains several different cell types and separate compartments and undergoes substantial development during its growth and maturation. Extracellular matrix (ECM) could be expected to play a major role in these processes. Most research on ECM in follicles has focused on the follicular basal lamina and its changing composition during folliculogenesis and on the specialised matrix formed at ovulation by the cumulus cells surrounding the oocyte and the zona pellucida. We review these aspects. Few naturally occurring gene mutations have identified unique roles for ECM molecules in follicular function. Presumably, any mutations leading to reduced fertility are eliminated quickly by natural selection and, when mutations are not eliminated, considerable redundancy occurs to ensure successful reproduction. In mice, in which the genome can be easily manipulated, the modification of matrix components associated with cumulus and oocytes has often resulted in partial infertility, suggesting redundancy. We provide an update of basal lamina components focusing on newer discoveries. In addition, we review matrix associated with the occyte and cumulus cells (excluding the zona pellucida) and other components of ECM. Where possible, we examine evidence for the role of the ECM in follicular development and diseases.
Collapse
Affiliation(s)
- Helen F Irving-Rodgers
- Research Centre for Reproductive Health, Department of Obstetrics and Gynaecology, University of Adelaide, 5005 Adelaide, Australia.
| | | |
Collapse
|
50
|
Eppig JJ, Pendola FL, Wigglesworth K, Pendola JK. Mouse Oocytes Regulate Metabolic Cooperativity Between Granulosa Cells and Oocytes: Amino Acid Transport1. Biol Reprod 2005; 73:351-7. [PMID: 15843493 DOI: 10.1095/biolreprod.105.041798] [Citation(s) in RCA: 176] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
A search for genes expressed more highly in mouse cumulus cells than mural granulosa cells by subtraction hybridization yielded Slc38a3. SLC38A3 is a sodium-coupled neutral amino acid transporter having substrate preference for l-glutamate, l-histidine, and l-alanine. Detectable levels of Slc38a3 mRNA were found by in situ hybridization in granulosa cells of large preantral follicles, but levels were higher in all granulosa cells of small antral follicles; expression became limited to cumulus cells of large antral follicles. Expression of Slc38a3 mRNA in granulosa cells was promoted by fully grown oocytes from antral follicles but not by growing oocytes from preantral follicles. Fully grown oocytes were dependent on cumulus cells for uptake of l-alanine and l-histidine but not l-leucine. Fully grown but not growing oocytes secreted one or more paracrine factors that promoted cumulus cell uptake of all three amino acids but of l-alanine and l-histidine to a much greater extent than l-leucine. Uptake of l-leucine appeared dependent primarily on contact-mediated signals from fully grown oocytes. Fully grown oocytes also promoted elevated levels of Slc38a3 mRNA and l-alanine transport by preantral granulosa cells, but growing oocytes did not. Therefore, fully grown oocytes secrete one or more paracrine factors that promote cumulus cell uptake of amino acids that oocytes themselves transport poorly. These amino acids are likely transferred to oocytes via gap junctions. Thus, oocytes use paracrine signals to promote their own development via metabolic cooperativity with cumulus cells. The ability of oocytes to mediate this cooperativity is developmentally regulated and acquired only in later stages of oocyte development.
Collapse
Affiliation(s)
- John J Eppig
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA.
| | | | | | | |
Collapse
|